Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Autoimmun ; 93: 131-138, 2018 09.
Article in English | MEDLINE | ID: mdl-30061015

ABSTRACT

The Foxa1 and Foxa2 transcription factors are essential for mouse development. Here we show that they are expressed in thymic epithelial cells (TEC) where they regulate TEC development and function, with important consequences for T-cell development. TEC are essential for T-cell differentiation, lineage decisions and repertoire selection. Conditional deletion of Foxa1 and Foxa2 from murine TEC led to a smaller thymus with a greater proportion of TEC and a greater ratio of medullary to cortical TEC. Cell-surface MHCI expression was increased on cortical TEC in the conditional Foxa1Foxa2 knockout thymus, and MHCII expression was reduced on both cortical and medullary TEC populations. These changes in TEC differentiation and MHC expression led to a significant reduction in thymocyte numbers, reduced positive selection of CD4+CD8+ cells to the CD4 lineage, and increased CD8 cell differentiation. Conditional deletion of Foxa1 and Foxa2 from TEC also caused an increase in the medullary TEC population, and increased expression of Aire, but lower cell surface MHCII expression on Aire-expressing mTEC, and increased production of regulatory T-cells. Thus, Foxa1 and Foxa2 in TEC promote positive selection of CD4SP T-cells and modulate regulatory T-cell production and activity, of importance to autoimmunity.


Subject(s)
Epithelial Cells/immunology , Hepatocyte Nuclear Factor 3-alpha/immunology , Hepatocyte Nuclear Factor 3-beta/immunology , T-Lymphocytes, Regulatory/immunology , Thymocytes/immunology , Thymus Gland/immunology , Animals , Autoimmunity , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Epithelial Cells/cytology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression Regulation , Hepatocyte Nuclear Factor 3-alpha/deficiency , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-beta/deficiency , Hepatocyte Nuclear Factor 3-beta/genetics , Lymphocyte Activation , Lymphocyte Count , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Organ Size , Signal Transduction , T-Lymphocytes, Regulatory/cytology , Thymocytes/cytology , Thymus Gland/cytology , Transcription Factors/genetics , Transcription Factors/immunology , AIRE Protein
2.
Development ; 145(3)2018 01 31.
Article in English | MEDLINE | ID: mdl-29361554

ABSTRACT

Gli3 is a Hedgehog (Hh)-responsive transcription factor that can function as a transcriptional repressor or activator. We show that Gli3 activity in mouse thymic epithelial cells (TECs) promotes positive selection and differentiation from CD4+ CD8+ to CD4+ CD8- single-positive (SP4) cells in the fetal thymus and that Gli3 represses Shh Constitutive deletion of Gli3, and conditional deletion of Gli3 from TECs, reduced differentiation to SP4, whereas conditional deletion of Gli3 from thymocytes did not. Conditional deletion of Shh from TECs increased differentiation to SP4, and expression of Shh was upregulated in the Gli3-deficient thymus. Use of a transgenic Hh reporter showed that the Hh pathway was active in thymocytes, and increased in the Gli3-deficient fetal thymus. Neutralisation of endogenous Hh proteins in the Gli3-/- thymus restored SP4 differentiation, indicating that Gli3 in TECs promotes SP4 differentiation by repression of Shh Transcriptome analysis showed that Hh-mediated transcription was increased whereas TCR-mediated transcription was decreased in Gli3-/- thymocytes compared with wild type.


Subject(s)
Hedgehog Proteins/metabolism , Nerve Tissue Proteins/metabolism , Thymocytes/cytology , Thymocytes/metabolism , Zinc Finger Protein Gli3/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Epithelial Cells/cytology , Female , Gene Expression Profiling , Hedgehog Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Pregnancy , Repressor Proteins/deficiency , Repressor Proteins/genetics , Repressor Proteins/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymocytes/immunology , Thymus Gland/cytology , Thymus Gland/embryology , Thymus Gland/metabolism , Zinc Finger Protein Gli3/deficiency , Zinc Finger Protein Gli3/genetics
3.
J Exp Med ; 214(7): 2041-2058, 2017 Jul 03.
Article in English | MEDLINE | ID: mdl-28533268

ABSTRACT

Before birth, B cells develop in the fetal liver (FL). In this study, we show that Gli3 activity in the FL stroma is required for B cell development. In the Gli3-deficient FL, B cell development was reduced at multiple stages, whereas the Sonic hedgehog (Hh [Shh])-deficient FL showed increased B cell development, and Gli3 functioned to repress Shh transcription. Use of a transgenic Hh-reporter mouse showed that Shh signals directly to developing B cells and that Hh pathway activation was increased in developing B cells from Gli3-deficient FLs. RNA sequencing confirmed that Hh-mediated transcription is increased in B-lineage cells from Gli3-deficient FL and showed that these cells expressed reduced levels of B-lineage transcription factors and B cell receptor (BCR)/pre-BCR-signaling genes. Expression of the master regulators of B cell development Ebf1 and Pax5 was reduced in developing B cells from Gli3-deficient FL but increased in Shh-deficient FL, and in vitro Shh treatment or neutralization reduced or increased their expression, respectively.


Subject(s)
B-Lymphocytes/metabolism , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Kruppel-Like Transcription Factors/genetics , Liver/metabolism , Nerve Tissue Proteins/genetics , Animals , Cell Lineage/genetics , Flow Cytometry , Gene Expression Profiling/methods , Liver/embryology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , PAX5 Transcription Factor/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Trans-Activators/genetics , Zinc Finger Protein Gli3
4.
Oncotarget ; 8(15): 24163-24176, 2017 Apr 11.
Article in English | MEDLINE | ID: mdl-28445929

ABSTRACT

Kif7 is a ciliary kinesin motor protein that regulates mammalian Hedgehog pathway activation through influencing structure of the primary cilium. Here we show that Kif7 is required for normal T-cell development, despite the fact that T-cells lack primary cilia. Analysis of Kif7-deficient thymus showed that Kif7-deficiency increases the early CD44+CD25+CD4-CD8- thymocyte progenitor population but reduces differentiation to CD4+CD8+ double positive (DP) cell. At the transition from DP to mature T-cell, Kif7-deficiency selectively delayed maturation to the CD8 lineage. Expression of CD5, which correlates with TCR signal strength, was reduced on DP and mature CD4 and CD8 cells, as a result of thymocyte-intrinsic Kif7-deficiency, and Kif7-deficient T-cells from radiation chimeras activated less efficiently when stimulated with anti-CD3 and anti-CD28 in vitro. Kif7-deficient thymocytes showed higher expression of the Hedgehog target gene Ptch1 than WT, but were less sensitive to treatment with recombinant Shh, and Kif7-deficient T-cell development was refractory to neutralisation of endogenous Hh proteins, indicating that Kif7-deficient thymocytes were unable to interpret changes in the Hedgehog signal. In addition, Kif7-deficiency reduced cell-surface MHCII expression on thymic epithelial cells.


Subject(s)
Cell Differentiation/genetics , Epithelial Cells/metabolism , Kinesins/genetics , Major Histocompatibility Complex/genetics , Thymocytes/cytology , Thymocytes/metabolism , Thymus Gland/physiology , Animals , Biomarkers , Gene Expression , Genotype , Hedgehog Proteins/metabolism , Major Histocompatibility Complex/immunology , Mice , Mice, Knockout , Phenotype , Signal Transduction , Thymocytes/immunology
5.
J Dev Biol ; 4(3): 22, 2016 Jul 16.
Article in English | MEDLINE | ID: mdl-27504268

ABSTRACT

T cells develop in the thymus, which provides an essential environment for T cell fate specification, and for the differentiation of multipotent progenitor cells into major histocompatibility complex (MHC)-restricted, non-autoreactive T cells. Here we review the role of the Hedgehog signalling pathway in T cell development, thymic epithelial cell (TEC) development, and thymocyte-TEC cross-talk in the embryonic mouse thymus during the last week of gestation.

6.
J Autoimmun ; 68: 86-97, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26778835

ABSTRACT

Sonic Hedgehog (Shh) is expressed in the thymus, where it regulates T cell development. Here we investigated the influence of Shh on thymic epithelial cell (TEC) development. Components of the Hedgehog (Hh) signalling pathway were expressed by TEC, and use of a Gli Binding Site-green fluorescence protein (GFP) transgenic reporter mouse demonstrated active Hh-dependent transcription in TEC in the foetal and adult thymus. Analysis of Shh-deficient foetal thymus organ cultures (FTOC) showed that Shh is required for normal TEC differentiation. Shh-deficient foetal thymus contained fewer TEC than wild type (WT), the proportion of medullary TEC was reduced relative to cortical TEC, and cell surface expression of MHC Class II molecules was increased on both cortical and medullary TEC populations. In contrast, the Gli3-deficient thymus, which shows increased Hh-dependent transcription in thymic stroma, had increased numbers of TEC, but decreased cell surface expression of MHC Class II molecules on both cortical and medullary TEC. Neutralisation of endogenous Hh proteins in WT FTOC led to a reduction in TEC numbers, and in the proportion of mature Aire-expressing medullary TEC, but an increase in cell surface expression of MHC Class II molecules on medullary TEC. Likewise, conditional deletion of Shh from TEC in the adult thymus resulted in alterations in TEC differentiation and consequent changes in T cell development. TEC numbers, and the proportion of mature Aire-expressing medullary TEC were reduced, and cell surface expression of MHC Class II molecules on medullary TEC was increased. Differentiation of mature CD4 and CD8 single positive thymocytes was increased, demonstrating the regulatory role of Shh production by TEC on T cell development. Treatment of human thymus explants with recombinant Shh or neutralising anti-Shh antibody indicated that the Hedgehog pathway is also involved in regulation of differentiation from DP to mature SP T cells in the human thymus.


Subject(s)
Cell Differentiation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Hedgehog Proteins/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Hedgehog Proteins/genetics , Humans , Mice , Mice, Knockout , Mice, Transgenic , Signal Transduction , Thymocytes/cytology , Thymocytes/immunology , Thymocytes/metabolism , Thymus Gland/immunology
7.
Oncotarget ; 6(30): 28646-60, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26415229

ABSTRACT

Developing thymocytes require pre-TCR signalling to differentiate from CD4-CD8- double negative to CD4+CD8+ double positive cell. Here we followed the transcriptional response to pre-TCR signalling in a synchronised population of differentiating double negative thymocytes. This time series analysis revealed a complex transcriptional response, in which thousands of genes were up and down-regulated before changes in cell surface phenotype were detected. Genome-wide measurement of RNA degradation of individual genes showed great heterogeneity in the rate of degradation between different genes. We therefore used time course expression and degradation data and a genome wide transcriptional modelling (GWTM) strategy to model the transcriptional response of genes up-regulated on pre-TCR signal transduction. This analysis revealed five major temporally distinct transcriptional activities that up regulate transcription through time, whereas down-regulation of expression occurred in three waves. Our model thus placed known regulators in a temporal perspective, and in addition identified novel candidate regulators of thymocyte differentiation.


Subject(s)
Cell Differentiation , Models, Genetic , Protein Precursors/genetics , Receptors, Antigen, T-Cell/genetics , Thymocytes/metabolism , Transcription, Genetic , Animals , Cells, Cultured , Cluster Analysis , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Genetic Markers , Genome-Wide Association Study , Genotype , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Homeodomain Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Phenotype , Protein Precursors/immunology , Protein Precursors/metabolism , RNA/genetics , RNA/metabolism , RNA Stability , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Thymocytes/immunology , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism
8.
J Cell Sci ; 128(11): 2085-95, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25908851

ABSTRACT

Different tissues contain diverse and dynamic cellular niches, providing distinct signals to tissue-resident or migratory infiltrating immune cells. Hedgehog (Hh) proteins are secreted inter-cellular signalling molecules, which are essential during development and are important in cancer, post-natal tissue homeostasis and repair. Hh signalling mediated by the Hh-responsive transcription factor Gli2 also has multiple roles in T-lymphocyte development and differentiation. Here, we investigate the function of Gli2 in T-cell signalling and activation. Gene transcription driven by the Gli2 transcriptional activator isoform (Gli2A) attenuated T-cell activation and proliferation following T-cell receptor (TCR) stimulation. Expression of Gli2A in T-cells altered gene expression profiles, impaired the TCR-induced Ca(2+) flux and nuclear expression of NFAT2, suppressed upregulation of molecules essential for activation, and attenuated signalling pathways upstream of the AP-1 and NFκB complexes, leading to reduced activation of these important transcription factors. Inhibition of physiological Hh-dependent transcription increased NFκB activity upon TCR ligation. These data are important for understanding the molecular mechanisms of immunomodulation, particularly in tissues where Hh proteins or other Gli-activating ligands such as TGFß are upregulated, including during inflammation, tissue damage and repair, and in tumour microenvironments.


Subject(s)
Kruppel-Like Transcription Factors/genetics , NF-kappa B/genetics , Receptors, Antigen, T-Cell/genetics , Signal Transduction/genetics , Transcription Factor AP-1/genetics , Transcriptional Activation/genetics , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Gene Expression Regulation/genetics , Hedgehog Proteins/genetics , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , NFATC Transcription Factors/genetics , T-Lymphocytes/metabolism , Transcriptome/genetics , Transforming Growth Factor beta/genetics , Up-Regulation/genetics , Zinc Finger Protein Gli2
9.
J Immunol ; 190(6): 2641-9, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23408837

ABSTRACT

Genome-wide association studies of complex immune-mediated diseases have indicated that many genetic factors, each with individual low risk, contribute to overall disease. It is therefore timely and important to characterize how immune responses may be subtly modified by tissue context. In this article, we explore the role of tissue-derived molecules in influencing the function of T cells, which, owing to their migratory nature, come into contact with many different microenvironments through their lifespan. Hedgehog (Hh) proteins act as secreted morphogens, providing concentration-dependent positional and temporal cell-fate specification in solid tissues. Hh signaling is required for embryogenesis and is important in postnatal tissue renewal and in malignancy. However, the function of Hh in dynamic, fluid systems, such as in mammalian immunity, is largely unknown. In this article, we show that Hh-dependent transcription in T cells promoted Th2 transcriptional programs and differentiation, exacerbating allergic disease. Of interest, expression of Sonic Hh increased in lung epithelial cells following the induction of allergic disease, and lung T cells upregulated Hh target gene expression, indicating that T cells respond to locally secreted Hh ligands in vivo. We show that Il4, the key Th2 cytokine, is a novel transcriptional target of Hh signals in T cells, providing one mechanism for the role of Hh in Th differentiation. We propose that Hh, secreted from inflamed, remodeling, or malignant tissue, can modulate local T cell function. Our data present an unexpected and novel role for tissue-derived morphogens in the regulation of fluid immune responses, with implications for allergy and tumor responses, suggesting new uses for anti-Hh therapeutics.


Subject(s)
Asthma/immunology , Asthma/metabolism , Cell Differentiation/immunology , Hedgehog Proteins/physiology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Animals , Asthma/pathology , Cells, Cultured , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/immunology , T-Lymphocytes, Helper-Inducer/pathology , Tissue Distribution/immunology , Transcription, Genetic/immunology
10.
J Invest Dermatol ; 133(5): 1221-30, 2013 May.
Article in English | MEDLINE | ID: mdl-23303453

ABSTRACT

Here we present a mouse model for T-cell targeting of hair follicles, linking the pathogenesis of alopecia to that of depigmentation disorders. Clinically, thymus transplantation has been successfully used to treat T-cell immunodeficiency in congenital athymia, but is associated with autoimmunity. We established a mouse model of thymus transplantation by subcutaneously implanting human thymus tissue into athymic C57BL/6 nude mice. These xenografts supported mouse T-cell development. Surprisingly, we did not detect multiorgan autoimmune disease. However, in all transplanted mice, we noted a striking depigmentation and loss of hair follicles. Transfer of T cells from transplanted nudes to syngeneic black-coated RAG(-/-) recipients caused progressive, persistent coat-hair whitening, which preceded patchy hair loss in depigmented areas. Further transfer experiments revealed that these phenomena could be induced by CD4+ T cells alone. Immunofluorescent analysis suggested that Trp2+ melanocyte-lineage cells were decreased in depigmented hair follicles, and pathogenic T cells upregulated activation markers when exposed to C57BL/6 melanocytes in vitro, suggesting that these T cells are not tolerant to self-melanocyte antigens. Our data raise interesting questions about the mechanisms underlying tissue-specific tolerance to skin antigens.


Subject(s)
Alopecia/physiopathology , CD4-Positive T-Lymphocytes/pathology , Cell Proliferation , Hair Color/physiology , Pigmentation/physiology , Thymus Gland/transplantation , Transplantation, Heterologous , Adoptive Transfer , Alopecia/pathology , Animals , CD4-Positive T-Lymphocytes/physiology , Disease Models, Animal , Hair Follicle/pathology , Hair Follicle/physiology , Homeodomain Proteins/genetics , Humans , In Vitro Techniques , Melanocytes/pathology , Melanocytes/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Up-Regulation/physiology
11.
Blood ; 119(20): 4741-51, 2012 May 17.
Article in English | MEDLINE | ID: mdl-22461491

ABSTRACT

The function of Hedgehog signaling in hematopoiesis is controversial, with different experimental systems giving opposing results. Here we examined the role of Desert Hedgehog (Dhh) in the regulation of murine erythropoiesis. Dhh is one of 3 mammalian Hedgehog family proteins. Dhh is essential for testis development and Schwann cell function. We show, by analysis of Dhh-deficient mice, that Dhh negatively regulates multiple stages of erythrocyte differentiation. In Dhh-deficient bone marrow, the common myeloid progenitor (CMP) population was increased, but differentiation from CMP to granulocyte/macrophage progenitor was decreased, and the mature granulocyte population was decreased, compared with wild-type (WT). In contrast, differentiation from CMP to megakaryocyte/erythrocyte progenitor was increased, and the megakaryocyte/erythrocyte progenitor population was increased. In addition, we found that erythroblast populations were Dhh-responsive in vitro and ex vivo and that Dhh negatively regulated erythroblast differentiation. In Dhh-deficient spleen and bone marrow, BFU-Es and erythroblast populations were increased compared with WT. During recovery of hematopoiesis after irradiation, and under conditions of stress-induced erythropoiesis, erythrocyte differentiation was accelerated in both spleen and bone marrow of Dhh-deficient mice compared with WT.


Subject(s)
Erythropoiesis/genetics , Hedgehog Proteins/physiology , Stress, Physiological/physiology , Age Factors , Animals , Bone Marrow/metabolism , Bone Marrow/radiation effects , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Differentiation/radiation effects , Cells, Cultured , Erythroblasts/metabolism , Erythroblasts/physiology , Erythroblasts/radiation effects , Erythropoiesis/physiology , Erythropoiesis/radiation effects , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Recovery of Function/genetics , Recovery of Function/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Signal Transduction/radiation effects , Spleen/metabolism , Spleen/radiation effects , Stress, Physiological/genetics , Stress, Physiological/radiation effects , Whole-Body Irradiation
12.
Eur J Immunol ; 42(2): 489-99, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22101858

ABSTRACT

In the thymus, developing T cells receive signals that determine lineage choice, specificity, MHC restriction and tolerance to self-antigen. One way in which thymocytes receive instruction is by secretion of Sonic hedgehog (Shh) from thymic epithelial cells. We have previously shown that Hedgehog (Hh) signalling in the thymus decreases the CD4:CD8 single-positive (SP) thymocyte ratio. Here, we present data indicating that double-positive (DP) thymocytes are Hh-responsive and that thymocyte-intrinsic Hh signalling plays a role in modulating the production of CD4(+) (SP4), CD8(+) (SP8) and unconventional T-cell subsets. Repression of physiological Hh signalling in thymocytes altered the proportions of DP and SP4 cells. Thymocyte-intrinsic Hh-dependent transcription also attenuated both the production of mature SP4 and SP8 cells, and the establishment of peripheral T-cell compartments in TCR-transgenic mice. Additionally, stimulation or withdrawal of Hh signals in the WT foetal thymus impaired or enhanced upregulation of the CD4 lineage-specific transcription factor Gata3 respectively. These data together suggest that Hh signalling may play a role in influencing the later stages of thymocyte development.


Subject(s)
Epithelial Cells/metabolism , Hedgehog Proteins/metabolism , T-Lymphocyte Subsets/metabolism , Thymocytes/metabolism , Thymus Gland/cytology , Animals , CD4 Antigens/genetics , CD4 Antigens/metabolism , CD8 Antigens/genetics , CD8 Antigens/metabolism , Cell Differentiation , Cell Lineage , Cells, Cultured , Embryo, Mammalian , Epithelial Cells/cytology , Epithelial Cells/immunology , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Hedgehog Proteins/genetics , Hedgehog Proteins/immunology , Mice , Mice, Knockout , Mice, Transgenic , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Thymocytes/cytology , Thymocytes/immunology , Thymus Gland/embryology , Thymus Gland/growth & development , Transcriptional Activation/genetics
13.
Cell Cycle ; 9(20): 4144-52, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20935514

ABSTRACT

The Hedgehog (Hh) signaling pathway influences multiple stages of murine T-cell development. Hh signaling mediates transcriptional changes by the activity of the Gli family of transcription factors, Gli1, Gli2 and Gli3. Both Gli2 and Gli3 are essential for mouse development and can be processed to function as transcriptional repressors or transcriptional activators, whereas Gli1, itself a transcriptional target of Hh pathway activation, can only function as a transcriptional activator and is not essential for mouse development. Gli1-deficient mice are healthy and appear normal and nonredundant functions for Gli1 have been difficult to identify. Here we show that Gli1 is non-redundant in the regulation of T-cell development in the thymus, at multiple developmental stages. Analysis of Gli1-deficient embryonic mouse thymus shows a role for Gli1 to promote the differentiation of CD4⁻CD8⁻ double negative (DN) thymocytes before pre- TCR signal transduction, and a negative regulatory function after pre-TCR signaling. In addition, introduction of a Class I-restricted transgenic TCR into the adult Gli1-deficient and embryonic Gli2-deficient thymus showed that both Gli1 and Gli2 influence its selection to the CD8 lineage.


Subject(s)
Cell Differentiation/physiology , Kruppel-Like Transcription Factors/metabolism , Signal Transduction/physiology , T-Lymphocytes/physiology , Thymus Gland/cytology , Thymus Gland/embryology , Thymus Gland/growth & development , Animals , Biomarkers/metabolism , Female , Hedgehog Proteins/metabolism , Kruppel-Like Transcription Factors/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Thymus Gland/metabolism , Zinc Finger Protein GLI1
SELECTION OF CITATIONS
SEARCH DETAIL
...